Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Cell Biol ; 96(5): 1475-9, 1983 May.
Article in English | MEDLINE | ID: mdl-6302102

ABSTRACT

We used affinity chromatography to isolate a specific laminin-binding protein from murine fibrosarcoma cells. These cells bind exogenous laminin to their surface with high affinity (Kd = 2 X 10(-9)M for laminin) with approximately 5 X 10(4) sites per cell. Laminin affinity chromatography of [35S]methionine-labeled cell extracts produced two distinct proteins. One was identified as Type IV (basement membrane) collagen based on its migration pattern on SDS gels and bacterial collagenase sensitivity. The other protein, which migrates as a single band or closely spaced doublet on reduced SDS gels, has a reduced molecular weight of 69,000. Using a nitrocellulose filter disk assay, we found that the latter protein specifically bound 125I-laminin with the same high affinity (Kd = 2 X 10(-9)M for laminin) as did intact fibrosarcoma cells. By iodinating intact cells, we demonstrated that this laminin-binding protein is on the cell surface. We conclude that this protein with reduced molecular weight of 69,000 is a subunit or component of a larger cell surface receptor protein for laminin in this fibrosarcoma model. This laminin receptor may mediate the interaction of the cell with its extracellular matrix.


Subject(s)
Fibrosarcoma/analysis , Glycoproteins/metabolism , Receptors, Cell Surface/isolation & purification , Animals , Chromatography, Affinity , Fibrosarcoma/metabolism , Kinetics , Laminin , Mice , Molecular Weight , Receptors, Laminin
2.
Science ; 244(4912): 1585-7, 1989 Jun 30.
Article in English | MEDLINE | ID: mdl-2662405

ABSTRACT

The growth of human mammary cells may be regulated by a balance between growth stimulatory and growth inhibitory pathways. Polypeptides of 47 and 65 kilodaltons (mammastatin) were isolated from conditioned medium of normal human mammary cells. Monoclonal antibodies against mammastatin were generated that blocked its activity and were used for purification and further characterization of the protein. Mammastatin inhibited the growth of 5 transformed human mammary cell lines, but had no effect on the growth of 11 transformed human cell lines derived from nonmammary tissues. Mammastatin appeared to be a heat-labile protein distinct from transforming growth factor-beta (TGF-beta). By immunoperoxidase staining it was detected in cultured normal human mammary cells, but was decreased in transformed mammary cells.


Subject(s)
Breast/metabolism , Growth Inhibitors/biosynthesis , Peptide Biosynthesis , Antibodies, Monoclonal , Breast/analysis , Breast/cytology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division , Cell Line, Transformed , Chromatography , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Epithelium/metabolism , Female , Growth Inhibitors/isolation & purification , Growth Inhibitors/pharmacology , Hot Temperature , Humans , Immunoassay , Immunoenzyme Techniques , Intercellular Signaling Peptides and Proteins , Molecular Weight , Peptides/isolation & purification , Peptides/pharmacology , Trypsin/pharmacology , Tumor Cells, Cultured
3.
Clin Pharmacol Ther ; 81(6): 893-8, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17460605

ABSTRACT

As cancer stem cells (SCs) drive tumor growth, it is only through the elimination of those cancer SCs that a pharmacologic cure can be attained. To study ways to develop drugs that target cancer SC, we investigated changes in cellular mechanisms and kinetics that occur in SC populations during colorectal cancer (CRC) development. We used computer modeling to determine which changes could give rise to exponential increases in both SC and non-SC populations in CRC. Our results show that the only mechanism that can explain how these subpopulations increase exponentially in CRC development involves an increase in symmetric SC cell division. This finding suggests that any systemic therapies designed to effectively treat CRC and other cancers must act to control or eliminate symmetrical cancer SC division in tumors, while minimally affecting normal SC division in non-tumor tissues.


Subject(s)
Cell Division , Colorectal Neoplasms/pathology , Models, Biological , Neoplastic Stem Cells/pathology , Colorectal Neoplasms/metabolism , Computer Simulation , Humans , Neoplastic Stem Cells/metabolism
4.
J Clin Invest ; 75(6): 2085-90, 1985 Jun.
Article in English | MEDLINE | ID: mdl-4008654

ABSTRACT

We have developed a long-term culture system in which murine marrow cells are cultured on a complex extracellular matrix (ECM) that is derived from marrow and extracted with guanidine hydrochloride and dithiothreitol. Marrow cultures were established with fresh murine marrow cells and recharged at 2 wk (week 0). Phase microscopy showed a dramatically increased adherent cell layer development on ECM compared with controls within a week after recharge. By electron microscopy, this adherent layer was composed of numerous reticular cells apparently attached to the ECM which extended cytoplasmic projections to the surrounding hematopoietic cells. Adherent cellularity on ECM-coated dishes increased to 30 times the control values by week 2. Cumulative suspension cells on ECM dishes were eight times controls. ECM influenced both hematopoietic progenitor cell proliferation and differentiation. Adherent colony-forming unit-granulocyte/macrophage and colony-forming unit-megakaryocyte were greater than 30 and 15 times the control values, respectively, by week 2 (P less than or equal to 0.05). There were more mature granulocytic and megakaryocytic cells in ECM-coated dishes than in controls at all time points. This new culture system directly demonstrates that ECM is an important component of the hematopoietic microenvironment.


Subject(s)
Bone Marrow Cells , Extracellular Matrix/physiology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Animals , Cell Adhesion , Cell Differentiation , Cell Division , Cells, Cultured , Granulocytes/cytology , Male , Mice , Rabbits
5.
J Clin Invest ; 75(3): 970-5, 1985 Mar.
Article in English | MEDLINE | ID: mdl-3980732

ABSTRACT

Long-term production of murine hematopoietic cells in vitro is dependent on establishment of a complex microenvironment consisting of a variety of stromal cells and an extensive extracellular matrix which includes collagen, fibronectin, laminin, proteoglycans, and other undefined components adherent to the culture dishes. Cis-4-hydroxyproline (CHP), a relatively specific inhibitor of collagen secretion, was used to examine the role of extracellular collagen deposition in supporting hematopoiesis in long-term C57B1/6J mouse bone marrow cell cultures. Throughout the 10-wk culture period, all culture dishes contained either 0, 10, 25, or 50 micrograms/ml of CHP. All medium and nonadherent cells were removed at weekly intervals and replaced with fresh medium containing the previous concentrations of CHP. Nonadherent cells were assayed weekly for total cells and pluripotent, erythroid, megakaryocytic, and granulocytic-macrophage progenitor cells. Dishes were killed at selected intervals to assess protein and collagen synthesis in the adherent layer. Adherent cell numbers, as judged by microscopic examination and DNA assays, correlated inversely with CHP concentrations used and paralleled degree of collagen synthesis inhibition. The decreased hemopoietic progenitor cell production correlated closely with percent inhibition of collagen synthesis and stromal cellularity. The CHP concentrations tested were not directly toxic to hemopoietic progenitor cells. These studies demonstrate that collagen deposition in the extracellular matrix of murine bone marrow cell cultures is essential to the establishment of a functional stromal microenvironment that is supportive of long-term hematopoiesis.


Subject(s)
Bone Marrow Cells , Collagen/metabolism , Extracellular Matrix/metabolism , Hematopoiesis/drug effects , Hydroxyproline/pharmacology , Animals , Cell Division/drug effects , Collagen/biosynthesis , Colony-Forming Units Assay , Extracellular Matrix/drug effects , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred C57BL
6.
Oncogene ; 36(42): 5793-5807, 2017 10 19.
Article in English | MEDLINE | ID: mdl-28604744

ABSTRACT

Previous studies have suggested that overexpression of the oncogenic protein epithelial membrane protein-2 (EMP2) correlates with endometrial carcinoma progression and ultimately poor survival from disease. To understand the role of EMP2 in the etiology of disease, gene analysis was performed to show transcripts that are reciprocally regulated by EMP2 levels. In particular, EMP2 expression correlates with and helps regulate the expression of several cancer stem cell associated markers including aldehyde dehydrogenase 1 (ALDH1). ALDH expression significantly promotes tumor initiation and correlates with the levels of EMP2 expression in both patient samples and tumor cell lines. As therapy against cancer stem cells in endometrial cancer is lacking, the ability of anti-EMP2 IgG1 therapy to reduce primary and secondary tumor formation using xenograft HEC1A models was determined. Anti-EMP2 IgG1 reduced the expression and activity of ALDH and correspondingly reduced both primary and secondary tumor load. Our results collectively suggest that anti-EMP2 therapy may be a novel method of reducing endometrial cancer stem cells.


Subject(s)
Endometrial Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Immunoglobulin G/pharmacology , Isoenzymes/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Neoplastic Stem Cells/metabolism , Retinal Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Female , Gene Expression Profiling , Humans , Isoenzymes/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Retinal Dehydrogenase/genetics , Xenograft Model Antitumor Assays
7.
J Natl Cancer Inst ; 75(2): 353-9, 1985 Aug.
Article in English | MEDLINE | ID: mdl-3860688

ABSTRACT

Proline analogues such as cis-4-hydroxy-L-proline (CHP) and L-azetidine-2-carboxylic acid (A2C) were tested for their antitumor activity in tissue culture and in vivo. In culture, CHP specifically inhibited those tumor cells that synthesized basement-membrane collagen. CHP appeared to selectively inhibit collagen biosynthesis with only a slight effect on protein synthesis. Culturing cells on type IV collagen matrix did not alter the antiproliferative effect of CHP. The inhibition of 450.1 mouse mammary tumor cells was fully reversible when cultures were incubated for 6 or 12 hours with 25 micrograms CHP/ml but was irreversible after 24 hours of exposure. Of the proline analogues tested against 450.1 tumor cells, A2C and CHP were the most potent inhibitors of cell growth. These two compounds were therefore tested in vivo using 3 transplantable tumors, all of which synthesized basement-membrane collagen. CHP and A2C were given twice daily to mice for 7 to 10 days at doses ranging from 50 mg/kg (body wt) to 600 mg/kg (body wt) per injection. Both CHP and A2C were completely inactive against the 450.1 mammary tumor and the EHS sarcoma. Both compounds also caused considerable liver toxicity. Against CD8F1 mammary tumors, treatment with maximum tolerated doses of CHP and A2C resulted in a slight but insignificant inhibition of tumor growth. While our studies confirmed previous findings that CHP specifically inhibited those tumor cells that synthesized basement-membrane collagen, CHP and A2C did not appear to be efficacious antitumor agents.


Subject(s)
Antimetabolites, Antineoplastic , Collagen/biosynthesis , Proline/analogs & derivatives , Animals , Azetidinecarboxylic Acid/pharmacology , Basement Membrane/metabolism , Cell Division/drug effects , Cell Line , Humans , Hydroxyproline/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , Proline/pharmacology , Sarcoma, Experimental/drug therapy
8.
Cancer Res ; 38(11 Pt 2): 4091-100, 1978 Nov.
Article in English | MEDLINE | ID: mdl-100216

ABSTRACT

A cell line, the growth and survival of which is markedly affected by linoleic acid, has been established from a carcinogen-induced rat mammary tumor. The cells have been continuously passaged in 5% rat serum plus 10% fetal calf serum-supplemented medium. The rat serum component was found to be indispensalbe, for when it was omitted the growth rate rapidly declined and the cells died by 5 to 7 days. Removal of the rat serum from the growth medium also resulted in a dramatic loss of Oil Red O-positive droplets in the cells, suggesting that the lipid component of rat serum might be a major growth-promoting principle in rat serum. This is likely since the total lipid fraction, but not the delipidized protein fraction, could largely supplant requirement of the cells for rat serum. Pure linoleic acid was found to be effective in maintaining the cell growth in delipidized serum or in whole fetal calf serum-supplemented medium. Fatty acid analysis revealed a 19-fold higher amount of linoleic acid in rat serum than in fetal calf serum.


Subject(s)
Fatty Acids, Unsaturated/pharmacology , Mammary Neoplasms, Experimental/drug therapy , 9,10-Dimethyl-1,2-benzanthracene , Animals , Cell Division/drug effects , Cell Line , Cell Survival/drug effects , Culture Media , Fatty Acids, Unsaturated/administration & dosage , Female , Linoleic Acids/pharmacology , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/ultrastructure , Microscopy, Electron , Rats
9.
Cancer Res ; 41(7): 2855-62, 1981 Jul.
Article in English | MEDLINE | ID: mdl-7018671

ABSTRACT

The growth of primary N-nitrosomethylurea-induced rat mammary tumors was depressed by cis-hydroxyproline (CHP). This growth arrest appeared to be related to the ability of CHP to inhibit the deposition of basement membrane collagen as based on the following observations: (a) in vitro and in vivo, tumor cells synthesized type IV collagen, the collagen uniquely localized in basement membranes; (b) in vitro, the inhibition of tumor cell growth was preceded by a specific decrease in collagen accumulation with no effect on non-collagen protein synthesis; (c) a transplantable N-nitrosomethylurea-induced rat mammary tumor accumulated no type IV collagen as determined by polyacrylamide gel electrophoresis and indirect immunofluorescence. The growth of this tumor was not influenced by CHP; (d) an established human mammary tumor cell line, MCF-7, did not accumulate type IV collagen and was not inhibited by CHP. At the doses which effectively blocked the growth of primary N-nitrosomethylurea-induced mammary tumors, CHP and no toxic effects, and serum prolactin levels were not altered. The inhibitory effect was thus apparently due to the direct action of CHP upon the accumulation of collagen in cells which required type IV collagen production for continued growth.


Subject(s)
Hydroxyproline/pharmacology , Mammary Neoplasms, Experimental/chemically induced , Methylnitrosourea , Nitrosourea Compounds , Animals , Autoradiography , Basement Membrane/metabolism , Breast Neoplasms/metabolism , Cell Line , Collagen/biosynthesis , Female , Fluorescent Antibody Technique , Humans , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Proline/pharmacology , Protein Biosynthesis , Rats
10.
Cancer Res ; 56(9): 1965-9, 1996 May 01.
Article in English | MEDLINE | ID: mdl-8616832

ABSTRACT

Bcl-xs is a dominant negative repressor of Bcl-2 and Bcl-xL, both of which inhibit apoptosis. We used a replication-deficient adenoviral vector to transiently overexpress Bcl-xs in MCF-7 human breast cancer cells, which overexpress Bcl-xL. Infection with this vector induced apoptosis in vitro. We then determined the effects of intratumoral injection of bcl-xs adenovirus on solid MCF-7 tumors in nude mice. Tumors injected four times with the bcl-xs adenovirus showed a 50% reduction in size. Using terminal transferase-mediated dUTP-digoxigenin nick end labeling, we observed apoptotic cells at sites of bcl-xs adenoviral injection. These experiments demonstrate the feasibility of using bcl-xs gene therapy to induce apoptosis in human breast tumors.


Subject(s)
Apoptosis/genetics , Genetic Therapy , Mammary Neoplasms, Experimental/therapy , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/genetics , Animals , Female , Gene Transfer Techniques , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Neoplasm Transplantation , bcl-X Protein
11.
Cancer Res ; 55(12): 2507-10, 1995 Jun 15.
Article in English | MEDLINE | ID: mdl-7780958

ABSTRACT

Resistance to apoptosis plays an important role in tumors that are refractory to chemotherapy. We report that Bcl-XL, which functions like Bcl-2 to inhibit apoptosis, is highly expressed in MCF-7 human breast carcinoma cells. We used Bcl-XS, a dominant negative inhibitor of Bcl-2 and Bcl-XL, to demonstrate the role of these genes in modulating chemotherapy-induced apoptosis. Bcl-XS overexpressed in MCF-7 cells by stable transfection does not affect viability by itself but induces a marked increase in chemosensitivity to VP-16 or taxol. Using an ELISA assay which quantitates DNA damage, we demonstrate that this sensitization is due to apoptosis, suggesting the therapeutic utility of targeting this pathway.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Paclitaxel/toxicity , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/biosynthesis , Repressor Proteins , Breast Neoplasms , Cell Division/drug effects , Cell Line , Enzyme-Linked Immunosorbent Assay , Humans , Kanamycin Kinase , Oncogene Proteins, Viral/metabolism , Papillomaviridae/genetics , Papillomaviridae/metabolism , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Recombinant Proteins/biosynthesis , Transfection , Tumor Cells, Cultured , bcl-X Protein
12.
J Clin Oncol ; 18(15): 2805-10, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10920127

ABSTRACT

PURPOSE: Medical care for clinical trials is often not reimbursed by insurers, primarily because of concern that medical care as part of clinical trials is expensive and not part of standard medical practice. In June 2000, President Clinton ordered Medicare to reimburse for medical care expenses incurred as part of cancer clinical trials, although many private insurers are concerned about the expense of this effort. To inform this policy debate, the costs and charges of care for patients on clinical trials are being evaluated. In this Association of American Cancer Institutes (AACI) Clinical Trials Costs and Charges pilot study, we describe the results and operational considerations of one of the first completed multisite economic analyses of clinical trials. METHODS: Our pilot effort included assessment of total direct medical charges for 6 months of care for 35 case patients who received care on phase II clinical trials and for 35 matched controls (based on age, sex, disease, stage, and treatment period) at five AACI member cancer centers. Charge data were obtained for hospital and ancillary services from automated claims files at individual study institutions. The analyses were based on the perspective of a third-party payer. RESULTS: The mean age of the phase II clinical trial patients was 58.3 years versus 57.3 years for control patients. The study population included persons with cancer of the breast (n = 24), lung (n = 18), colon (n = 16), prostate (n = 4), and lymphoma (n = 8). The ratio of male-to-female patients was 3:4, with greater than 75% of patients having stage III to IV disease. Total mean charges for treatment from the time of study enrollment through 6 months were similar: $57,542 for clinical trial patients and $63,721 for control patients (1998 US$; P =.4) CONCLUSION: Multisite economic analyses of oncology clinical trials are in progress. Strategies that are not likely to overburden data managers and clinicians are possible to devise. However, these studies require careful planning and coordination among cancer center directors, finance department personnel, economists, and health services researchers.


Subject(s)
Clinical Trials as Topic/economics , Health Care Costs/statistics & numerical data , Insurance, Health, Reimbursement , Medicare/economics , Neoplasms/therapy , Aged , Cost-Benefit Analysis , Data Collection , Female , Humans , Male , Middle Aged , Neoplasms/economics , Pilot Projects , Public Policy , United States
13.
Oncogene ; 34(6): 671-80, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-24531711

ABSTRACT

Somatic mutations or deletions of TP53 and PTEN in ductal carcinoma in situ lesions have been implicated in progression to invasive ductal carcinomas. A recent molecular and mutational analysis of breast cancers revealed that inactivation of tumor suppressors, p53 and PTEN, are strongly associated with triple negative breast cancer. In addition, these tumor suppressors have important roles in regulating self-renewal in normal and malignant stem cells. To investigate their role in breast carcinogenesis, we knocked down these genes in human mammary cells and in non-transformed MCF10A cells. p53 and PTEN knockdown synergized to activate pro-inflammatory interleukin-6 (IL6)/Stat3/nuclear factor κB signaling. This resulted in generation of highly metastatic epithelial-to-mesenchymal transition-like cancer stem cells resulting in tumors whose gene expression profile mimicked that found in basal/claudin-low molecular subtype within the triple negative breast tumors. Constitutive activation of this loop in transformed cells was dependent on proteolytic degradation of suppressor of cytokine signaling 3 (SOCS3) resulting in low levels of this protein in basal/claudin-low cell lines and primary tumors. In non-transformed cells, transient activation of the IL6 inflammatory loop induced SOCS3 expression leading to pathway inactivation. In transformed cells, enforced expression of SOCS3 or interfering with IL6 pathway via IL6R blockade inhibited tumor growth and metastasis in mouse xenograft models. Furthermore, circulating tumor cells were significantly reduced in tumor-bearing animals when treated with anti-IL6R antibodies. These studies uncover important connections between inflammation and carcinogenesis and suggest that blocking pro-inflammatory cytokines may be utilized as an attractive strategy to target triple negative breast tumors, which currently lacks molecularly targeted therapies.


Subject(s)
Carcinoma, Intraductal, Noninfiltrating/genetics , PTEN Phosphohydrolase/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Animals , Carcinogenesis , Carcinoma, Intraductal, Noninfiltrating/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Inflammation/genetics , Inflammation/pathology , Interleukin-6/metabolism , Mice , Receptors, Interleukin-6/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
14.
Hum Gene Ther ; 9(8): 1209-16, 1998 May 20.
Article in English | MEDLINE | ID: mdl-9625260

ABSTRACT

Replication-deficient viral vectors are currently being used in gene transfer strategies to treat cancer cells. Unfortunately, viruses are limited in their ability to diffuse through tissue. This makes it virtually impossible to infect the majority of tumor cells in vivo and results in inadequate gene transfer. This problem can be addressed by allowing limited viral replication. Limited viral replication facilitates greater penetration of virions into tissue and can improve gene transfer. We have developed a strategy of limited viral replication using AdRSVlaclys, a chemically modified E1-deleted adenovirus, to codeliver an exogenous plasmid encoding the adenovirus E1 region. This system allows one round of viral replication. We examined the effect of this limited adenovirus replication in vitro and in vivo. In culture, codelivery of virus and pE1 resulted in a large increase in infected cells when compared with control cells exposed to virus and pUC19. In experiments on nude mice bearing HeLa ascites tumors, intraperitoneal injection of AdRSVlaclys/pE1 resulted in a significantly higher percentage of infected HeLa cells as compared with the PBS controls (p < 0.05) or the AdRSVlaclys/pUC19 controls (p < 0.01). These data demonstrate that the transcomplementation of replication-deficient adenovirus with exogenous E1 DNA leads to limited replication, and this controlled replication enhances gene transfer efficiency of adenovirus in vivo.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Neoplasms, Experimental/therapy , Virus Replication , Animals , Genetic Complementation Test , HeLa Cells , Humans , Lac Operon , Mice , Mice, Nude , Neoplasms, Experimental/pathology , Plasmids/genetics
15.
Hum Gene Ther ; 11(14): 2009-24, 2000 Sep 20.
Article in English | MEDLINE | ID: mdl-11020800

ABSTRACT

The efficiency of gene therapy strategies against cancer is limited by the poor distribution of the vectors in the malignant tissues. To solve this problem, a new generation of tumor-specific, conditionally replicative adenoviruses is being developed. To direct the replication of the virus to breast cancer, we have considered one characteristic present in a great proportion of these cancers, which is the expression of estrogen receptors (ERs). On the basis of the wild-type adenovirus type 5, we have constructed a conditionally replicative adenovirus (Ad5ERE2) in which the E1a and E4 promoters have been replaced by a portion of the pS2 promoter containing two estrogen-responsive elements (EREs). This promoter induces transcriptional activation of the E1a and E4 units in response to estrogens in cells that express the ERs. Ad5ERE2 is able to kill ER(+) human breast cancer cell lines as efficiently as the wild-type virus, but has decreased capacity to affect ER(-) cells. By complementation of the E1a protein in trans, Ad5ERE2 allows restricted replication of a conventional E1a-deleted adenoviral vector. When a virus expressing the proapoptotic gene Bc1-xs (Clarke et al., Proc. Natl. Acad. Sci. U.S.A. 1995;92:11024-11028) is used in combination with Ad5ERE2, the ability of both viruses to induce cell death is dramatically increased, and the effect can be modulated by addition of the antiestrogen tamoxifen.


Subject(s)
Adenoviridae/genetics , Adenovirus E1A Proteins/genetics , Breast Neoplasms/therapy , Genetic Therapy/methods , Genetic Vectors , Adenovirus E4 Proteins/genetics , Animals , Cell Death , Estrogens/genetics , Estrogens/pharmacology , Female , Gene Deletion , Genes, Reporter , Humans , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Genetic , Plasmids/metabolism , Promoter Regions, Genetic , Receptors, Estrogen/genetics , Response Elements/genetics , Tamoxifen/pharmacology , Time Factors , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Cells, Cultured
16.
Neoplasia ; 2(3): 251-60, 2000.
Article in English | MEDLINE | ID: mdl-10935511

ABSTRACT

Oncogenes which drive the cell cycle, such as c-myc, can sensitize cells to apoptosis. This suggests the possibility that the expression of genes such as bcl-2 or bcl-xL is required to inhibit apoptosis induced by oncogene expression. We hypothesized that inhibition of Bcl-2/Bcl-xL by the pro-apoptotic Bcl-xS protein, would result in selective induction of apoptosis in mammary carcinoma cells compared to their nontransformed counterparts. Therefore, we compared the effects of Bcl-xS expression delivered by a bcl-xS adenovirus (bcl-xS-Adv) vector, on viability and apoptosis of nontransformed versus transformed mammary epithelial cells. We report that c-myc-transformed murine mammary cells are extremely sensitive to apoptosis induced by the bcl-xS adenovirus (bcl-xS-Adv) vector, whereas immortalized, nontransformed murine mammary cells are relatively resistant to apoptosis induced by this vector. Likewise, human mammary epithelial cells transduced with c-erbB-2 were more sensitive to apoptosis induced by the bcl-xS vector than the nontransformed parental cells. Similar results were obtained when we tested the effects of bcl-xS adenoviral infection on primary normal human mammary epithelial cells and SUM-190 PT cells, (a c-erbB-2 over-expressing human mammary carcinoma cell line) grown on Matrigel. These data are consistent with the hypothesis that inhibition of Bcl-2/Bcl-xL can result in selective killing of cancer cells compared to their nontransformed counterparts.


Subject(s)
Adenoviridae/genetics , Apoptosis , Mammary Glands, Animal/cytology , Mammary Neoplasms, Experimental/therapy , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Cell Line , Cell Transformation, Neoplastic , Fluorouracil/pharmacology , Genes, myc , Humans , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Receptor, ErbB-2/physiology , bcl-X Protein
17.
Cancer Epidemiol Biomarkers Prev ; 9(3): 257-63, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10750663

ABSTRACT

We examined the effects of all-trans retinoic acid (ATRA) and lutein (a nonprovitamin A carotenoid), on apoptosis and chemosensitivity in primary normal human mammary epithelial cells, SV40 transformed mammary cells, and MCF-7 human mammary carcinoma cells. ATRA and lutein selectively induced apoptosis in transformed but not normal human mammary cells. In addition, both compounds protected normal cells, but not transformed cells, from apoptosis induced by the chemotherapy agents etoposide and cisplatin. Furthermore, lutein and ATRA selectively increased the ratio of Bcl-xL:Bax protein expression in normal cells but not transformed mammary cells, suggesting a possible mechanism for selective modulation of apoptosis. The differential effects of lutein and ATRA on apoptotic pathways in normal versus transformed mammary epithelial cells may have important implications for chemoprevention and therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Lutein/pharmacology , Tretinoin/pharmacology , Chemoprevention , Female , Gene Expression Regulation, Neoplastic , Genes, p53/genetics , Humans , Simian virus 40/pathogenicity , Tumor Cells, Cultured
18.
Invest Ophthalmol Vis Sci ; 21(6): 879-82, 1981 Dec.
Article in English | MEDLINE | ID: mdl-7031010

ABSTRACT

Dog corneas were studied by indirect immunofluorescence with antibodies specific for pemphigus and pemphigoid antigens, laminin, and type IV collagen. The distribution of these antigens was similar to that observed in other squamous epithelia and skin. This study provides further evidence for close immunochemical similarities between these tissues.


Subject(s)
Antigens/immunology , Cornea/immunology , Pemphigoid, Bullous/immunology , Pemphigus/immunology , Skin Diseases, Vesiculobullous/immunology , Animals , Collagen/immunology , Dogs , Epithelium/immunology , Fluorescent Antibody Technique , Glycoproteins/immunology , Laminin
19.
Cancer Lett ; 12(1-2): 9-21, 1981 Mar.
Article in English | MEDLINE | ID: mdl-6791817

ABSTRACT

7,12-Dimethylbenzanthracene-induced rat mammary tumors contain basement membrane collagen in vivo and in vitro in primary culture, as determined by immunofluorescence. The proline analogue dis-hydroxyproline, which selectively blocks collagen accumulation, inhibits tumor cell growth in vitro. In vivo cis-hydroxyproline blocks tumor growth and produces epithelial cell degeneration and tumor necrosis at a non-toxic dose. This compound has no effect on the growth of the mammary carcinoma cell line 64/24, which makes no basement membrane collagen. As we have previously described for the normal proliferating rat mammary gland, basement membrane collagen deposition may be necessary for the growth of some rat mammary tumors.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene , Basement Membrane/metabolism , Benz(a)Anthracenes , Collagen/metabolism , Mammary Neoplasms, Experimental/metabolism , Animals , Collagen/antagonists & inhibitors , Female , Hydroxyproline/pharmacology , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Rats
20.
Biochem Pharmacol ; 58(6): 1067-74, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10509759

ABSTRACT

Didemnin B (DB) is one member of a class of natural cyclic depsipeptides that display potent cytotoxicity in vitro. The detailed mechanism of action of DB is unknown, although it appears to involve the inhibition of protein biosynthesis. Additional activities of DB have established DB as a rapid and potent inducer of apoptosis in HL-60 cells. Our aim was to determine if the induction of apoptosis by DB is mediated through inhibition of protein synthesis in MCF-7 human breast carcinoma cells. Apoptosis was observed only at > or = 100 nM DB, even though inhibition of protein synthesis occurred at much lower DB concentrations (IC50 = 12 nM). DB-induced apoptosis was mediated by caspase activation, since cleavage of the caspase substrate poly(ADPribose) polymerase was observed as early as 6 hr after DB exposure. Two additional protein synthesis inhibitors, cycloheximide (CHX) and emetine (ET), failed to induce apoptosis at concentrations that completely inhibited protein synthesis. Moreover, DB-induced apoptosis was enhanced only slightly by pre- and co-treatment with CHX and ET. Thus, inhibition of protein synthesis alone was not sufficient to induce apoptosis in these cells. As a measure of antiproliferative potential, DB (1-5 nM) inhibited the colony forming ability of MCF7 cells regardless of pretreatment with CHX. In conclusion, additional effects of DB, independent of protein synthesis inhibition, are proposed to account for its ability to induce apoptosis and prevent cell proliferation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Depsipeptides , Peptides, Cyclic/pharmacology , Protein Synthesis Inhibitors/pharmacology , Breast Neoplasms , Dose-Response Relationship, Drug , Humans , Poly(ADP-ribose) Polymerases/metabolism , Stem Cells/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL